Accepted Manuscript Title: Progress Report on New Antiepileptic Drugs: A Summary of the Twelfth Eilat Conference (EILAT XII) Author: Meir Bialer Svein I. Johannessen Ren´e H. Levy Emilio Perucca Torbj¨orn Tomson H. Steve White PII: DOI: Reference:

S0920-1211(15)00004-2 http://dx.doi.org/doi:10.1016/j.eplepsyres.2015.01.001 EPIRES 5307

To appear in:

Epilepsy Research

Received date: Accepted date:

10-12-2014 9-1-2015

Please cite this article as: Bialer, M., Johannessen, S.I., Levy, R.H., Perucca, E., Tomson, T., White, H.S.,Progress Report on New Antiepileptic Drugs: A Summary of the Twelfth Eilat Conference (EILAT XII), Epilepsy Research (2015), http://dx.doi.org/10.1016/j.eplepsyres.2015.01.001 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Progress Report on New Antiepileptic Drugs: A Summary of the Twelfth Eilat Conference (EILAT XII) Meir Bialera,*, Svein I. Johannessenb, René H. Levyc, Emilio Peruccad, Torbjörn

cr

a

ip t

Tomsone, H. Steve Whitef

Institute for Drug Research, School of Pharmacy and David R. Bloom Center for

us

Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel

The National Center for Epilepsy, Sandvika, and Department of Pharmacology, Oslo

an

b

University Hospital, Oslo, Norway

Departments of Pharmaceutics and Neurological Surgery, University of Washington,

Seattle, Washington, WA, USA

Division of Clinical and Experimental Pharmacology, Department of Internal

d

d

M

c

te

Medicine and Therapeutics, University of Pavia, and C. Mondino National

Ac ce p

Neurological Institute, Pavia, Italy e

Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden

f

Department of Pharmacology and Toxicology, University of Utah, Salt Lake City,

UT, USA

1 Page 1 of 166

*Correspondence: Prof. Meir Bialer Institute for Drug Research, School of Pharmacy and David R. Bloom Centre for

ip t

Pharmacy, Faculty of Medicine, Ein Karem The Hebrew University of Jerusalem,

cr

91120 Jerusalem, Israel tel: +972-2-6758610 fax +972-2-6757246

an

us

e-mail: [email protected]

Highlights

M

 The Twelfth Eilat Conference on New Antiepileptic Drugs (AEDs)-EILAT XII  Antiepileptic Drugs in development

 Preclinical evaluation and clinical trials

Ac ce p

te

d

 Development strategies of new AEDs

2 Page 2 of 166

Summary The Twelfth Eilat Conference on New Antiepileptic Drugs (AEDs)-EILAT XII, took

ip t

place in Madrid, Spain from August 31st to September 3rd, 2014. About 130 basic scientists, clinical pharmacologists and neurologists from 22 countries attended the

cr

conference, whose main themes included "Conquering pharmacoresistant epilepsy" ,

‘"Innovative emergency treatments", “Progress report on second-generation

us

treatment” and "New methods and formulations". Consistent with previous formats of this conference, a large part of the program was devoted to a review of AEDs in

an

development, as well as updates on AEDs introduced since 2004. Like the EILAT X and EILAT XI reports, the current article focuses on the preclinical and clinical

releasing

silk,

allopregnanolone

M

pharmacology of AEDs that are currently in development. These include adenosine(SAGE-547),

AMP-X-0079,

brivaracetam,

d

bumetanide, cannabidiol, cannabidivarin, 2-deoxy-glucose, everolimus, ganaxolone,

te

huperzine A, imepitoin, minocycline, NAX 801-2, pitolisant, PRX 0023, SAGE-217,

Ac ce p

valnoctamide and its homologue sec-propylbutylacetamide (SPD), and VLB-01. Since the previous Eilat conference, perampanel has been introduced into the market and twelve novel potential epilepsy treatments are presented for the first time.

Keywords: Antiepileptic drugs, Drug development, Epilepsy, Pharmacology, Clinical trials, Conference

3 Page 3 of 166

Adenosine-Releasing Silk

us

cr

ip t

D. Boison

an

Robert Stone Dow Neurobiology Labs, Legacy Research Institute, Portland, OR, USA

M

Adenosine

Introduction and rationale for development

d

Adenosine is an endogenous anticonvulsant responsible for seizure arrest and postictal

te

refractoriness (Boison, 2007). However, maladaptive changes in adenosine

Ac ce p

homeostasis occur during epileptogenesis and induce a deficiency in adenosine – a characteristic pathological hallmark of human temporal lobe epilepsy (Aronica et al., 2013). In addition, reduced adenosine drives epigenetic changes thought to be instrumental in epileptogenesis (Williams-Karnesky et al., 2013). Therefore, adenosine augmentation represents a rational approach to prevent seizures and the progression of epilepsy (Boison, 2012). Systemic, largely cardiovascular, side effects of adenosine require a local mode of adenosine delivery. To deliver a defined dose of adenosine with a known release kinetics locally to the brain, a silk biopolymer was used as drug delivery system. Purified silk fibroin presents a unique option for therapeutic adenosine delivery as it is biocompatible and biodegrades slowly. Degradation kinetics can be regulated to allow control of release from weeks to years. 4 Page 4 of 166

Both silk as well as adenosine are already FDA-approved for different indications and the frequent use of silk sutures in brain confirms the feasibility of implanting silk

ip t

biomaterials into brain.

Pharmacology

cr

Adenosine acts as endogenous ligand of four types of G protein coupled adenosine

receptors (A1, A2A, A2B, and A3) (Chen et al., 2013). In addition, adenosine provides

us

biochemical feedback inhibition of DNA methylation (Williams-Karnesky et al.,

an

2013). Adenosine homeostasis in the brain is largely under the control of metabolic clearance through adenosine kinase (ADK) expression in astrocytes (Boison, 2013).

M

Anticonvulsant profile

Demonstration of the therapeutic efficacy of adenosine augmentation therapy (AAT)

d

has been documented in at least 15 published research studies in two different species

te

(mice and rats), and in four different models of epilepsy (mouse intrahippocampal and intraamygdaloid kainic acid (KA) model; rat hippocampal kindling model; rat

Ac ce p

systemic KA model) (Boison, 2012). In all four models robust seizure suppression or prevention in the absence of discernible side effects was achieved by focal AAT mediated by: (i) intraventricular implants of adenosine releasing polymeric devices; (ii) intraventricular or infrahippocampal implants of cells engineered to release

adenosine; and (iii) AAV-based gene therapy designed to reduce ADK expression in

astrocytes using antisense technology. Therapeutic efficacy has been demonstrated in a combined total of >150 different experimental epileptic animals. Dose response studies have shown that intraventricular doses of 50 to 500 ng adenosine per kg body weight per day provide effective seizure suppression in rodents, whereas doses of up to 5000 ng adenosine per kg per day were without any discernible side effects. To 5 Page 5 of 166

circumvent systemic side effects of global adenosine augmentation, a local (epileptogenic region) therapeutic approach becomes a necessity to harness the powerful anti-seizure potential of adenosine in the absence of side effects. Local AAT

ip t

affords reliable bioavailability of adenosine within the epileptogenic brain region. AAT effectively suppressed seizures in three different models of epilepsy:

cr

Mouse intra-hippocampal KA model: Pharmacological augmentation of adenosine

seizures that were refractory to carbamazepine.

us

signaling (ADK inhibitor or adenosine A1R agonist) completely (0/1900sz) prevented

an

Mouse intra-amygdaloid KA model: Infra-hippocampal implants of adenosinereleasing mouse embryonic stem cell-derived neural precursor cells (releasing 20-40

M

ng adenosine per day) completely (0/400sz) prevented the development of seizures when cells were implanted 24 h after the SE and animals were assessed for

d

spontaneous seizures at 3 weeks.

te

Rat kindling model: Adenosine releasing brain implants (polymers, encapsulated cells, stem cell derived implants) solidly suppressed epileptic seizures and retarded

Ac ce p

kindling epileptogenesis. Those studies demonstrated that the local paracrine release of adenosine resulting in adenosine concentrations 2000 ng adenosine per

M

day) were well tolerated, without any gross behavioral changes of the animals. To study anti-epileptogenic properties of adenosine, we used silk-based polymers

d

designed to release 1000 ng adenosine per day during a limited time span of ten days.

te

When transplanted into the brain ventricle of fully kindled rats those implants completely suppressed kindled seizures only during the time span of active adenosine

Ac ce p

release. After expiration of adenosine release from the polymers seizures gradually resumed. When rats were kindled in the presence of adenosine releasing silk, seizure development was suppressed. Importantly, following a washout period of adenosine from the silk, kindling stimulations did not trigger any generalized seizures, indicating a novel anti-epileptogenic effect of focal adenosine delivery (Szybala et al., 2009). To further substantiate the anti-epileptogenic effect of transient adenosine release, we implanted silk-polymers releasing 250 ng adenosine / ventricle / day into the brain ventricles of epileptic rats 9 weeks after the systemic administration of KA. During active adenosine release (restricted to 10 days) the incidence of spontaneous recurrent seizures was markedly suppressed (by ~75%) in epileptic rats. Furthermore, this 7 Page 7 of 166

transient therapeutic intervention reversed the DNA hypermethylation seen in the epileptic brain, inhibited sprouting of mossy fibers in the hippocampus, and prevented the progression of epilepsy for at least 3 months. Thus, pathological changes in DNA

ip t

methylation homeostasis may underlie epileptogenesis and reversal of these epigenetic changes with AAT may halt disease progression longterm (Williams-

us

cr

Karnesky et al., 2013).

Other pharmacological properties

an

In addition to seizure suppression and prevention, adenosine augmentation has neuroprotective, antipsychotic and pro-cognitive properties (Boison, 2013; Shen et al.,

M

2012).

te

Anti-ictogenic mechanisms

d

Mechanism(s) of action

Binding of adenosine to pre- and postsynaptic A1 receptors (Ki: 77nM; high

Ac ce p

expression levels in the limbic system) inhibits adenylyl cyclase activity, activates potassium channels, blocks transient calcium channels and increases intracellular calcium and inositol-1,4,5-trisphosphate levels by activating phospholipase C. Through these mechanisms A1 receptors block transmitter release and reduce the neuronal firing rate (Chen et al., 2013). Anti-epileptogenic mechanisms Adenosine is an obligatory metabolic endproduct of transmethylation reactions, which include DNA methylation. Increased adenosine shifts the S-adenosylhomocysteine (SAH) hydrolase reaction towards increased SAH production, which inhibits DNA methyltransferases through product inhibition (Williams-Karnesky et al., 2013). 8 Page 8 of 166

Transient therapeutic adenosine augmentation reduces DNA methylation status of the epileptic rodent brain and prevents disease progression long term through this

ip t

epigenetic mechanism.

Toxicology

cr

Adenosine is an endogenous nucleoside that occurs in all cells of the body and is

subject to endogenous metabolic clearance mechanisms. Adenosine is FDA-approved

us

for the treatment of supraventricular tachycardia and available commercially in a

an

preservative-free formulation (3 mg / ml) for intravenous use. In addition, the safety of intrathecal (IT) adenosine in humans has been addressed: In a preclinical toxicity

M

screening no side effects were observed in dogs with IT adenosine at 10µl/kg/h (2.4 mg/day for 48 hours, then 7.2 mg/day for 26 days). In reports using IT adenosine

d

agonists for the treatment of neuropathic pain in humans, investigators demonstrated

te

statistically significant reduction of neuropathic pain and allodynia by IT injection of the adenosine agonist R-PIA. No adverse effects were reported. In a Phase I clinical

Ac ce p

safety study 1 mg adenosine was injected IT in 12 volunteers with neuropathic pain. No adverse effects were noticed. Additional Phase I studies demonstrated the general safety of intrathecal adenosine administration in concentrations of up to 2 mg.

Pharmacokinetics and metabolic profile Intravenously administered adenosine, in concentrations used for the treatment of supraventricular tachycardia in humans, is rapidly cleared from the circulation via cellular uptake, primarily by erythrocytes and vascular endothelial cells. Intracellular adenosine is rapidly metabolized either via phosphorylation to AMP by ADK, or via deamination to inosine by adenosine deaminase (ADA) in the cytosol (Boison, 2013). 9 Page 9 of 166

Since ADK has a lower Km and Vmax than ADA, deamination plays a significant role only when cytosolic adenosine saturates the phosphorylation pathway. Inosine formed by deamination of adenosine can leave the cell intact or can be degraded to

ip t

hypoxanthine, xanthine, and finally uric acid. AMP formed by phosphorylation of adenosine is incorporated into the high-energy phosphate pool. In the brain, adenosine

cr

clearance is largely mediated by ADK activity (Boison, 2013). Extracellular adenosine is primarily cleared by cellular uptake with a half-life of less than 10

us

seconds in whole blood and less than 10 minutes in brain. Excessive amounts of

an

adenosine can be deaminated by adenosine ecto-deaminase (ectoADA). Adenosine requires no hepatic or renal function for its activation or inactivation, and hepatic and

M

renal failure would not be expected to alter its effectiveness or tolerability.

d

Drug interactions

te

The methylxanthines theophylline and caffeine competitively antagonize adenosine's

Ac ce p

effects on adenosine receptors. Dipyridamole potentiates the action of adenosine.

Efficacy data

Efficacy data of adenosine releasing silk are available from rodent models of epilepsy as described above. It is estimated that the minimally effective dose of adenosine delivered by a local brain implant is in the range of 50 to 500 ng adenosine per kg per day.

Tolerability and adverse effect profile Adverse effects of intravenous adenosine (6 mg given as a rapid intravenous bolus administered over a 1 to 2 second period) in adult subjects for the treatment of 10 Page 10 of 166

supraventricular tachycardia have been found to affect a variety of systems. Cardiovascular effects included facial flushing (18%), headache (2%), sweating, palpitations, chest pain, hypotension (less than 1%). Respiratory effects included

ip t

shortness of breath/dyspnea (12%), chest pressure (7%), hyperventilation, head pressure (less than 1%). Central Nervous System (CNS) effects included

cr

lightheadedness (2%), dizziness, tingling in arms, numbness (1%), apprehension,

blurred vision, burning sensation, heaviness in arms, neck and back pain (less than

us

1%). Gastrointestinal effects were nausea (3%), metallic taste, tightness in throat,

an

pressure in groin (less than 1%). The local application of adenosine in the brain is

M

expected to avoid any of the above-mentioned systemic non-CNS side effects.

Planned studies

d

Further preclinical studies for the application “anti-epileptogenesis” are planned. This

te

includes determination of minimally effective anti-epileptogenic doses of adenosine, optimization of the delivery procedure, further mechanistic studies and long-term

Ac ce p

efficacy studies in rodent models of epileptogenesis.

11 Page 11 of 166

Allopregnanolone (SAGE-547) Injection S.J. Kanes, J. Doherty, R. Hammond, M. Rogawski, A. Robichaud, J. Jonas.

M

Allopregnanolone (SAGE-547)

an

us

cr

ip t

Sage Therapeutics, Cambridge, MA 02142, USA

d

Introduction and rationale for development

te

SAGE-547 (allopregnanolone) Injection is a solution of 5 mg/mL allopregnanolone

Ac ce p

and 250 mg/mL betadex sulfobutyl ether sodium, NF (Captisol®). Allopregnanolone Injection (SAGE-547) is being developed for the treatment of super-refractory SE (SRSE).

Allopregnanolone is an endogenously occurring neuroactive steroid and a principal metabolite of progesterone formed in the corpus luteum of the ovary, adrenal cortex and CNS (Paul & Purdy, 1992). Endogenous concentrations of allopregnanolone are at their highest in women during the third trimester of pregnancy, and approximate 157 nM at time of parturition (Luisi et al, 2000). Allopregnanolone is a potent positive allosteric modulator of GABAA receptor responses (Lambert et al,

2003). Through GABAA receptor modulation,

allopregnanolone possesses potent anxiolytic, sedative and anticonvulsant activity 12 Page 12 of 166

when studied in non-clinical in vitro and in vivo model systems (Belelli et al, 1989; Bitran et al, 1991; Wieland et al, 1991).

Anticonvulsant profile (animal models/electrophysiology)

ip t

Pharmacology

cr

There is ample evidence indicating a role for allopregnanolone in the treatment of seizures and status epilepticus (SE). Allopregnanolone has anticonvulsant properties

us

in a variety of acute seizure models, including the pentylenetetrazol (PTZ), 6Hz, The acute anticonvulsant efficacy of

an

bicuculline, and picrotoxin models.

allopregnanolone in the PTZ-induced seizure model in mice is maintained upon repeat

M

dosing. Allopregnanolone produces complete suppression of generalized amygdalakindled convulsions and protection against pilocarpine- or kainate-induced limbic

d

seizures and SE, with higher protective index values than clonazepam. In the rat

te

kainate-induced model of SE, allopregnanolone (30 mg/kg, ip) eliminated SE whether administered at either 10 minutes or at 70 minutes following kainate administration,

Ac ce p

whereas diazepam (5 mg/kg, ip) was only effective at 10 minutes following kainate administration.

These results support the hypothesis that enhancement of extra-

synaptic GABAA receptor function by allopregnanolone may provide anticonvulsant

efficacy when prolonged seizure activity has become pharmaco-resistant to benzodiazepine treatment.

Mechanism(s) of action Mammalian GABAA receptors are heteropentameric chloride-conducting ion channels that mediate fast inhibition of synaptic transmission via a reduction of neuronal membrane excitability. These ionotropic receptors are called GABAA receptors to 13 Page 13 of 166

distinguish them from metabotropic GABA receptor (G-protein coupled) (GABAB) receptors that mediate a slower form of synaptic inhibition. Allosteric potentiation of GABAA receptor function by allopregnanolone has been

ip t

extensively documented (Belelli, 2002; Belelli et al, 2005). Consistent with this literature, SAGE-547 potentiated GABA-mediated currents from recombinant human

cr

GABAA receptors expressed in heterologous mammalian cell lines. SAGE-547 produced a potent, concentration-dependent enhancement of GABA-evoked currents

us

recorded using whole-cell patch electrophysiology from Ltk-1 cells expressing α1β2γ2

an

receptors, with a half maximal effective concentration (EC50) of 60 nM and a maximal potentiation of 380% for agonist (EC20)-induced currents. SAGE-547 also produced a

M

potent enhancement of α4β3δ receptors, with an EC50 of 80 nM and a maximal potentiation of 418% for agonist (EC20)-induced currents.

d

In addition to allosteric enhancement of channel function, allopregnanolone

te

modulates the binding of a variety of other ligands to the GABAA receptor, including the picrotoxin/convulsant site labeled by [35S]-t-butylbicyclophosphorothionate

Ac ce p

(TBPS) (Concas et al, 1996). Consistent with this literature, SAGE-547 potently inhibited [35S]-TBPS binding, displacing the radioligand from rat cerebral cortex membranes with an inhibition constant (Ki) of 18 nM.

Toxicology

The minimum lethal doses for allopregnanolone in mice, rats, and rabbits are 20, 15, and 7 mg/kg intravenously, respectively. Based on the correction for body surface area across the species, these are equivalent to human doses of 1.6, 2.4, and 2.25 mg/kg intravenously, or 112, 168, and 158 mg in a 70 kg human (Gyermek et al, 1968). 14 Page 14 of 166

A recently published abstract presented the determination of maximum tolerated doses (MTD) of allopregnanolone in neonatal beagle dogs after bolus and i.v. infusions (Steinmetz et al, 2013). One pup administered allopregnanolone via i.v.

ip t

bolus (50 mg/kg), was not responsive to stimuli and exhibited, shallow rapid breathing but fully recovered without intervention. One pup administered 20 mg/kg

cr

allopregnanolone over a 4 hr i.v. infusion, fell asleep approximately 30 min post dose

and recovered without intervention. No allopregnanolone-related adverse effects on

us

chemistry or hematology labs were observed. Of note, studies with allopregnanolone

an

by the same investigators suggested that the presence of isoflurane anesthesia resulted in an MTD of 2-fold in one of the three donors. Although SAGE-547 shows the potential to induce CYP2B6, the increase was small

Ac ce p

at the highest concentration tested. Clinical exposures are targeted at 150 nM, which are well below the 30-50 μM required for induction of the enzyme. The potential for SAGE-547 (1 and 10 µM) to inhibit human CYP1A2, CYP2C9, CYP 2C19, CYP2D6, and CYP3A4 was investigated in vitro utilizing human liver

microsomes incubated in the presence of clinically relevant marker substrates. No inhibition was observed at 1 µM of SAGE-547. At 10 µM, no inhibition was observed for any of the CYPs, except for CYP2C9, which showed 47% inhibition. This result was followed up by determining half maximal inhibitory concentration (IC50) and Ki for CYP2C9 utilizing human liver microsomes and measuring formation of 4’hydroxy-diclofenac from diclofenac. IC50 was 0.41 µM and Ki was 0.256 µM. There 16 Page 16 of 166

was no evidence of time-dependent or metabolism-dependent inhibition based on monitoring IC50-shift after pre-incubation. SAGE-547 has the potential to alter the

ip t

metabolism of CYP2C9 substrates when administered concomitantly.

Tolerability and adverse effect profile

cr

With systemic exposures up to 150 nM, no drug-related serious TEAEs have been reported. Drug related TEAEs reported with i.v. allopregnanolone are generally mild,

us

the most frequently reported (>5% patients) being feelings of intoxication, sedation,

an

and vertigo. One patient experienced an anxiety attack, which is potentially a withdrawal effect. Clinical observations in the i.v. studies included decreased saccadic

M

eye movements, reduced episodic memory, as well as reduced plasma levels of LH and FSH. (Timby et al, 2006; Timby et al, 2011; van Broekhoven et al, 2007; Kask et

te

d

al, 2008; Kask et al, 2009).

Efficacy data and ongoing study

Ac ce p

An open-label Phase I/II clinical trial was initiated to study the safety, tolerability and

efficacy of SAGE-547 in at least 10 adult patients with SRSE. This trial is enrolling patients who have been in SE for at least 24 h despite treatment with first and second line SE treatments and have either failed to have SE controlled after 24 hours on a third-line general anesthetic (GA) agent or if SE was controlled by a GA have failed at least 1 wean attempt. A patient is excluded from the trial if pregnant or lactating, or if the patient has a known allergy to progesterone or allopregnanolone, has clinically significant ECG abnormalities, has a significant medical or surgical condition that may compromise vital organ systems, has been exposed to another experimental treatment within 30 days, is receiving a continuous 17 Page 17 of 166

i.v. AED (third-line agent) for seizure suppression or burst-suppression that will require greater than 24 h to wean, has enrolled in this trial previously, or if the SRSE was due to anoxic/hypoxic encephalopathy.

The figure below demonstrates the

ip t

design of the screening and treatment periods of this trial. Following the treatment period, there will be an acute two day follow-up period and an extended three-week

an

us

cr

follow-up period.

M

Figure 1: Trial design of SABE-547 in Super-refractory status epilepticus (SRSE)

d

The primary objective of this trial are to evaluate the safety and tolerability of SAGE-

te

547 injection in SRSE patients as assessed by monitoring treatment-emergent adverse

Ac ce p

events (TEAE), EEG, physical and neurological examinations, vital signs, clinical laboratory measures, ECGs and concomitant medication usage.

The secondary

objective of this trial is to assess the efficacy of SAGE-547 in SRSE, a assessed by the need to place the patient back into general anesthesia for seizure control during administration of SAGE-547 prior to taper.

Other secondary objectives include

duration of response, as well as changes in behavior as measured by rating scales of agitation, depth of coma, and survival. This trial is ongoing. To date, 4 patients have completed the full 5 day administration of SAGE-547 at a target plasma exposure of 150 nM and 30-day follow-up period. All have met the key efficacy endpoint of SE control during SAGE-547 administration. To date there have been no drug-related

18 Page 18 of 166

serious TEAEs in these patients.

Planned studies

Ac ce p

te

d

M

an

us

cr

larger scale controlled trial in a similar population of SRSE patients.

ip t

Depending upon the outcome of the Phase I/II trial, the plan is to follow-up with a

19 Page 19 of 166

AMP-X-0079 A. Pesyan1, R. J Porter2 AurimMed Pharma, Park City, Utah, USA1

cr

ip t

University of Pennsylvania, Philadelphia, PA, and USUHS, Bethesda MD, USA2

Introduction and rationale for development

us

AurimMed Pharma, Inc. is developing innovative prescription drugs using the Privileged Structure Platform™ (PSP) strategy to generate lead compounds. The PSP

an

approach ensures that the process of drug design proceeds exclusively from a highly enriched pool of pharmaceutically and medicinally relevant chemical structures,

M

greatly shortening product development times while enhancing clinical success (Muller, 2003; DeSimone et al., 2004; Costantino and Barlocco, 2006; Duarte et al.,

d

2007). The PSP strategy has been used in the development of novel anti-seizure drug

Ac ce p

te

candidates with potential superior efficacy and greatly diminished adverse effects.

Pharmacology

Anticonvulsant profile (animal models/electrophysiology) Based on studies in animal models, AurimMed Pharma’s most advanced AED candidate, AMP-X-0079, has a very broad spectrum, with the potential to become a superb anti-seizure drug for a variety of seizure types, including absence/petit mal, tonic-clonic and myoclonic seizures as well as status epilepticus. In addition to the

compound’s broad spectrum of activities across a number of animal models (Tables 1 and 2), it has (in rodent models) good oral bioavailability, CNS penetration, long duration of action, good neurological safety margins (Protective Indices, PIs), and substantial potency. 20 Page 20 of 166

AMP-X-0079 anticonvulsant data included in Tables 1 and 2 were largely derived from the Anticonvulsant Screening Program of NINDS, NIH. As one can see, AMPX-0079 is active in all the models tested, including the new semi-chronic mesial

ip t

temporal lobe epilepsy model. Protective Index (PI) = TD50/ED50. (“Toxicity” is defined here as rotarod motor impairment in mice and a battery of tests to determine

cr

neurological deficit as indicated by ataxia or other minimal motor impairment (MMI) in rats.)

us

In studies conducted by the NIH and the US Department of Defense (DOD), AMP-X-

an

0079 is active in models of chemically induced status epilepticus (SE), both when administered at the time of the chemical insult (time zero) as well as 30 minutes after

M

the appearance of seizures; the compound is therefore effective in both the prevention and the treatment of SE. Moreover, in a pilocarpine-induced model of SE, followed

d

by extensive, 9 day, evaluation in a Morris water maze task, AMP-X-0079 preserved

te

cognition, spatial learning and memory. The compound also demonstrated substantial in vivo neuroprotective effects in the dentate gyrus, CA1, and CA3 hippocampal

Ac ce p

neurons in the brains of a vast majority (87%) of the treated animals (results confirmed histologically). In the pilocarpine insult and SE, 100% of the AMP-X0079-treated animals survived compared to 54% of controls. In addition, the compound is active against non-convulsive seizures in benzodiazepine-resistant electrographic status epilepticus (ESE), confirmed by EEG studies (Lehmkuhle et al.,

2009).

AMP-X-0079 has been shown in in vivo studies to be orally bioavailable, fast-acting (rapid oral absorption within 15 minutes), with a reasonably long pharmacological half-life in animal models. The compound passes the blood-brain barrier, thereby manifesting its pharmacological effects directly in the central nervous system. 21 Page 21 of 166

Because of its unique pharmacological profile and significant suppression of ESE, the lead candidate (AMP-X-0079) was tested against a nerve agent (i.e., soman) by the U.S. Army Medical Research Institute of Chemical Defense (USAMRICD) of the

ip t

DOD. AMP-X-0079 is one of the few drugs capable of terminating ongoing status epilepticus seizures due to this nerve agent. It controls seizure very rapidly (typically

cr

50%, with a half-life from 2-8 h after oral administration. In vitro metabolic profiling was conducted in cryopreserved human, mouse, rat, and dog hepatocytes, and reveals essentially four discreet metabolites. Although structural identification of these metabolites has not been conducted to date,

109 Page 109 of 166

their molecular mass suggests that they all result from single hydroxylations. In addition, there is no evidence for the occurrence of unique human metabolites.

ip t

Drug interactions Although no formal drug interaction studies have been conducted, in vitro CYP

cr

inhibition (IC50) is >30 µM for all major isozymes, suggesting low potential for SAGE-217 to inhibit the metabolism of substrates for those enzymes. Induction of

us

drug metabolizing enzymes assessed in human hepatocytes indicates low level of

an

induction of CYP3A4 and CYP2B6, suggesting that SAGE-217 has the potential to

M

alter the pharmacokinetics of substrates for those enzymes.

Efficacy data

te

d

To date, no human studies have been completed with SAGE-217.

Tolerability and side effect profile

Ac ce p

To date, no human studies have been completed with SAGE-217.

Planned studies

Currently, , to support a first in man (FIM) study, a number of IND-enabling toxicology studies are planned or on-going, including 28-day general toxicology studies in rats and dogs, a genetic toxicity battery of Ames, rat micronucleus and Comet assays, and a safety pharmacology battery of rat CNS, rat respiratory and dog telemetry studies. In addition, to support later stage clinical investigations and a potential market launch, Sage Therapeutics will perform sub-chronic (3-month) and chronic (6 or 9-month) studies in rats and dogs, reproductive toxicology studies with 110 Page 110 of 166

Seg II in rats and rabbit. Rat Seg I and Seg III, a battery of abuse liability of drug dependence, drug discrimination and self administration, and carcinogenicity studies in rats and mice are also planned. Pending outcome of these trials, a Phase III clinical

Ac ce p

te

d

M

an

us

cr

ip t

program will be conducted.

111 Page 111 of 166

Valnoctamide and sec-Butylpropylacetamide (SPD): Second Generation Drugs to Valproic Acid (VPA)

1

ip t

M. Bialer,1 S. Kadosh2 and B.Yagen1 Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew

StatExcellence, Kfar Saba, Israel

us

2

cr

University of Jerusalem, Israel

CH3

CH2

an

CH3

CH3

CH2

CH

*

M

* CHCONH2 CH2

* CHCONH2 CH2

CH3

sec-Butyl-propylacetamide (SPD)

Ac ce p

te

Valnoctamide (VCD)

CH2

CH

*

d

CH3

CH3

Introduction and rationale for development Valnoctamide (VCD) is a CNS-active chiral constitutional isomer of valpromide, the corresponding amide of valproic acid (VPA). Since EILAT XI (2012), VCD underwent phase IIb clinical trial in patients with acute mania. VCD's one-carbon homologue sec-butyl-propylacetamide (SPD) was evaluated in a wide array of anticonvulsant models (White et al., 2012; Pouliot et al., 2013; Shekh-Ahmad et al., 2013). Both VCD and SPD possess two stereogenic centers in their chemical structure (denoted above with the asterisk *) and exhibit stereoselective pharmacokinetics (PK) in humans (VCD) and animals (Bialer and Yagen, 2007; Bialer, 2012). Consequently, the pharmacodynamics (PD; anticonvulsant activity) and PK of the four individual 112 Page 112 of 166

stereoisomers of VCD and SPD were evaluated in several rodent anticonvulsant models including: maximal electroshock (MES), 6Hz psychomotor, subcutaneous metrazol (scMet) and the pilocarpine- and soman-induced status epilepticus (SE) (Hen

ip t

et al., 2013; Shekh-Ahmad, 2014). The PK-PD (anticonvulsant activity) relationship of VCD and SPD stereoisomers were evaluated following i.p. administration (70 or 60

cr

mg/kg) to rats. Induction of neural tube defects (NTDs) by VCD and SPD stereoisomers was evaluated in a mouse strain highly-susceptible to teratogen-induced

an

us

NTD.

Pharmacology

M

Anticonvulsant profile (animal models/electrophysiology) Valnoctamide (VCD)

d

VCD (racemate and/or its individual enantiomers) demonstrated anticonvulsant

te

activity in a wide array of anticonvulsant models in mice and rats (MES, scMet and 6Hz, kindling) as described in the previous Eilat (EILAT X & XI) Conference

Ac ce p

manuscripts (Bialer et al., 2010 & 2013). In mice VCD four individual stereoisomers exhibited similar anticonvulsant activity

to one another as well as to racemic-VCD in the scMet and 6Hz tests, while at the MES test (2R,3R)-VCD was more potent than its enantiomer (2S,3S)-VCD and diastereoisomer (2S,3R)-VCD. In rats (p.o.) racemic-VCD and its four individual stereoisomers exhibited similar anticonvulsant activity in the MES test while (2R,3S)-VCD was the most potent compound at the scMet test with an ED50 value 5-

times more potent than the racemate (Shekh-Ahmad et al, 2014). Racemic-VCD as well as (2R,3S)-VCD and (2S,3S)-VCD administered at seizure onset blocked the pilocarpine-induced SE with similar ED50 values of about 40 113 Page 113 of 166

mg/kg. However, racemic-VCD and its individual strereoisomers lost this anti-SE activity when given (75-100 mg/kg) 30 min after seizure onset (Shekh-Ahmad et al., 2014).

ip t

Racemic-VCD administered with the standard medical countermeasures at treatment delays of 5 min, 20 min and 40 min after seizure onset was capable of stopping

cr

soman-induced SE seizures with ED50 values of 26mg/kg, 60mg/kg and 62mg/kg, respectively. Following administration of VCD the average latency (sec) for

us

electrographic seizure termination (mean±SEM) at 5 min, 20 min and 40 min was:

an

115±15, 497±15 and 1,336±318, respectively . VCD is one of the few drugs effective

sec-Butylpropylacetamide (SPD)

M

at 40 min delay at the soman-induced SE model (Shekh-Ahmad et al., 2014).

d

Racemic-SPD had a wide spectrum of anticonvulsant activity similar to VPA but was

te

4-30 times more potent than VPA in a wide array of anticonvulsant animal models (Shekh-Ahmad et al., 2013). Three SPD individual stereoisomers: (2R,3R)-SPD,

Ac ce p

(2S,3S)-SPD and (2R,3S)-SPD, exhibited anticonvulsant activity similar to racemicSPD, while (2S,3R)-SPD exhibited less potent anticonvulsant activity at the rat (po) MES model (Hen et al., 2013).

SPD stereoisomers, administered 30 min after the first observed Stage 3 motor seizure at the lithium-pilocarpine induced SE model, prevented the expression of further convulsive seizures in a dose-dependent fashion with ED50 values ranging

between 95-135 mg/kg. (2R,3R)-SPD was the least potent SPD stereoisomer (ED50 >130 mg/kg) (Hen et al., 2013). SPD and its individual stereoisomers were capable of stopping soman-induced SE seizures with ED50 values ranging between 40 to 71 mg/kg. Following administration 114 Page 114 of 166

of SPD individual stereoisomers the average latency (sec) for electrographic seizure termination at the 20-min treatment delay time (mean±SEM) was: 550±149; n=16 [(2R,3R)-SPD], 994±280; n=8 [(2R,3S)-SPD], 719±216; n=15 [(2S,3R)-SPD],

ip t

1589±684; n=13 [(2S,3S)-SPD] . All four individual SPD stereoisomers had a steep dose response curve with Hill (shape) coefficient of 4.5-8.9 that lead to a tight 95%

cr

confidence interval around their ED50 values. (2R,3R)-SPD was more potent than racemic-SPD as well as SPD three other individual stereoisomers (p

Progress report on new antiepileptic drugs: A summary of the Twelfth Eilat Conference (EILAT XII).

The Twelfth Eilat Conference on New Antiepileptic Drugs (AEDs) - EILAT XII, took place in Madrid, Spain from August 31st to September 3rd 2014. About ...
1MB Sizes 0 Downloads 16 Views